Eft226, a First in Class Inhibitor of Eif4a, Targets FGFR1/2 and HER2

Total Page:16

File Type:pdf, Size:1020Kb

Eft226, a First in Class Inhibitor of Eif4a, Targets FGFR1/2 and HER2 eFT226, a first in class inhibitor of eIF4A, targets FGFR1/2 and HER2 driven cancers B133 Peggy A. Thompson, Nathan P. Young, Craig R. Stumpf, Boreth Eam, Vikas K. Goel, Joan Chen, Sarah Fish, Gregory S. Parker, Adina Gerson-Gurwitz, Maria Barrera, Eric Sung, Jocelyn Staunton, Gary G. Chiang, Christopher J. Wegerski, Samuel Sperry, Kevin R. Webster, Siegfried H. Reich eFFECTOR Therapeutics, San Diego, CA Abstract Results Background: Mutations or amplifications affecting receptor tyrosine kinases (RTKs) activate eFT226 is a Sequence Selective Translational Regulator eFT226 Downregulates RTK Protein Expression and AKT/MAPK Signaling eFT226 Treatment Results in Tumor Growth Inhibition and Regression the RAS/MAPK and PI3K/AKT signaling pathways thereby promoting cancer cell MDA-MB-361 HER2 GAPDH proliferation and survival. Oncoprotein expression is tightly controlled at the level of mRNA A. B. A. A. MDA-MB-361 (BC: HER2amp) MDA-MB-361 (BC: HER2amp) translation and is regulated by the eukaryotic translation initiation factor 4F (eIF4F) HO N 3’ Tumor Volume Body Weight complex consisting of eIF4A, eIF4E, and eIF4G. eIF4A functions to catalyze the unwinding of O HO eIF4A1 A/G 60S secondary structure in the 5’-untranslated region (5’-UTR) of mRNA facilitating ribosome 40S scanning and translation initiation. eFT226 is a first in class inhibitor that converts eIF4A1 N eFT226 mRNA 80S Polysomes into a sequence specific translational repressor. eFT226 increases the affinity between O O A/G 1 2 3 4 5 6 7 8 9 10 eIF4A1 and polypurine recognition elements in the 5’-UTR leading to selective Fraction 5’ downregulation of mRNA translation. The polypurine element is highly enriched in the 5’- CN B. C. UTR of eFT226 target genes, many of which are known oncogenic drivers, including MDA-MB-361 (HER2amp) NCI-H716 (FGFR2amp) FGFR1/2 and HER2, enabling eFT226 to selectively inhibit dysregulated oncogene C. 0 10 30 100 eFT226 (nM) 0 10 25 50 100 eFT226 (nM) expression. HER2 FGFR2 Binding to AGAGAG RNA surface RNA KD (µM) KD (µM) Fold Methods: 5’-UTR dependency was evaluated using cell-based luciferase reporter assays. Sequence w/ eFT226 w/o eFT226 Change p-ERK p-ERK Regulation of protein expression was analyzed by western blot analysis. Antitumor activity AGAGAG 0.021 ±0.001 8.0 ±0.9 381 ERK ERK B. NCI-H716 (CRC: FGFR2amp) C. NCI-H1581 (NSCLC: FGFR1amp) was assessed in vitro by proliferation and apoptosis assays. For in vivo experiments, GGCGGC 3.19 ±0.03 8.0 ±0.3 2.5 p-AKT S473 p-AKT S473 Tumor Volume Tumor Volume athymic nude or NOD/SCID mice were implanted with subcutaneous xenograft models of CCGCCG 9.6 ±0.4 3.27 ±0.06 0.34 AKT AKT FGFR1, FGFR2 or HER2 driven tumors and treated with eFT226 administered Q4D IV. CAACAA 2.43 ±0.01 3.78 ±0.05 1.6 GAPDH Tubulin Results: eFT226 inhibits the translation of FGFR1, FGFR2 and HER2 through formation of a sequence dependent ternary complex with eIF4A1 and polypurine elements within the 5’- UTR of mRNA [eIF4A1-eFT226-mRNA]. Formation of this ternary complex blocks ribosome D. MDA-MB-361 E. NCI-H716 scanning along the 5’-UTR leading to dose dependent inhibition of RTK protein expression. Cells transiently transfected with luciferase reporter constructs containing the 5’-UTR of D. 5’-UTR eFT226 Rel. to Hipp. Rel. to each RTK resulted in 10-45-fold greater sensitivity to inhibition by eFT226 compared to a Sequence IC50 (nM) AGAGAG IC50 (nM) AGAGAG control 5’-UTR confirming the 5’-UTR dependency. In solid tumor cell lines driven by AGAGAG 1.5 ±0.4 1 426 1 alterations in FGFR1, FGFR2 or HER2, downregulation of RTK expression by eFT226 resulted GGCGGC 13.8 ±2.0 9.2 204 0.5 in decreased MAPK and AKT signaling, potent inhibition of cell proliferation and an CCGCCG 92 ±18 61.5 369 0.9 induction of apoptosis suggesting that eFT226 could be effective in treating tumor types CAACAA 218 ±55 145 210 0.5 D. dependent on these oncogenic drivers. Solid tumor xenograft models harboring FGFR1/2 F. MDA-MB-361 G. NCI-H716 or HER2 amplifications treated with eFT226 resulted in significant in vivo tumor growth inhibition and regression at well tolerated doses in breast, non-small cell lung and colorectal cancer models. Treatment with eFT226 also decreased RTK protein levels supporting the potential to use these eFT226 target genes as pharmacodynamic markers of Figure 1. eFT226 is a sequence selective translational regulator. A) Chemical structure of eFT226. B) Schematic of the ternary complex interactions [eIF4A-eFT226-mRNA]. C) eFT226 enhances the binding target engagement. of eIF4A1 to specific RNAs. A representative Biacore sensogram of eIF4A1 binding to an AGAGAG RNA Conclusions: eFT226 is efficacious against tumor models with alterations in FGFR1, FGFR2 surface in the presence (green) or absence (black) of eFT226 (left panel). The stability of ternary and HER2 RTKs. The antitumor response observed in preclinical in vivo models driven by complexes formed on different RNA sequences is summarized (right panel). D) Inhibition of in vitro H. Proliferation Apoptosis Apoptosis RTK amplifications demonstrates the potential for eFT226 in the treatment of solid tumors translation by eFT226 is sequence dependent. Luciferase reporter gene constructs containing 5’-UTRs Cell line Tumor type RTK with FGFR1/2 or HER2 alterations. Furthermore, this data provides a means to select with 6-mer sequence motif repeats were transiently transfected into the MDA-MB-231 cell line and GI50 (nM) IC50 (nM) Emax sensitive patient subsets during clinical development. Clinical trials in patients with solid treated with increasing concentrations of eFT226 or hippuristanol for 4 hr. Green: AGAGAG, Blue: BT474 Breast HER2 4.4 2 3 right panel MDAMB361 Breast HER2 11.9 16 8 tumor malignancies have initiated. GGCGGC, Red: CCGCCG and Black: CAACAA. IC50 values are summarized in the table ( ). MDAMB231 Breast FGFR1 10.9 15 10 Figure 5. eFT226 inhibits the growth of RTK driven xenografts in vivo. A) MDA-MB-361 (HER2amp) MFM223 Breast FGFR2 3.1 6 14 xenograft-bearing animals were treated Q4D with either vehicle or eFT226 IV (1 mg/kg or 0.1 mg/kg) for NCIH716 Colorectal FGFR2 8.4 23 4 28 days. eFT226 treatment results in significant tumor regression (left panel). eFT226 is well tolerated as NCIH1581 Non-small cell lung FGFR1 5.7 90 1.6 Introduction Inhibition of eFT226 Target Genes is Mediated Through the 5’-UTR seen by the body weight measurements taken throughout the tumor growth study (right panel). B) amp Figure 3. eFT226 translationally regulates key oncogenic drivers resulting in inhibition of growth and Treatment of NCI-H716 (FGFR2 ) xenograft-bearing animals with the indicated doses of eFT226 Q4D RTKs A. amp survival. A) Polysome profile of MDA-MB-361 cells treated with 20 nM eFT226 or control for 3 hr (left results in tumor growth inhibition and regression after 15 days. C) Treatment of NCI-H1581 (FGFR1 ) 5’-UTR eFT226 panel). eFT226 inhibits the translation of HER2 (center panel) but not GAPDH (right panel) as shown by xenograft-bearing animals with the indicated doses of eFT226 Q4D results in significant tumor growth • eIF4A RNA helicase is an essential Sequence IC (nM) 50 qPCR analysis of mRNA from polysome fractions. B-C) Treatment of MDA-MB-361 (B) or NCI-H716 (C) cell inhibition after 20 days. D) Tumor growth inhibition values at day 14-15 in RTK (FGFR1, FGFR2 and HER2) component of the translation FGFR1 0.8 driven xenograft models for the indicated doses of eFT226 (Q4D IV). Dotted line indicates 100% tumor PI3K RAS lines with eFT226 for 24 hr decreases RTK protein levels, AKT and MAPK signaling as assessed by western initiation complex that regulates FGFR2 1.7 blot analysis. D-E) RTK and MAPK or AKT signaling are co-regulated. F-G) Down regulation of RTK protein growth inhibition. PTEN RAF multiple key oncogenes involved in HER2 4.2 levels results in inhibition of cell growth and an induction of apoptosis. H) Tabulated summary of AKT TUBA 36 proliferation (measured by Cell Titer-Glo) and apoptosis (measured by Caspase-Glo 3/7) values for RTK MEK tumor cell proliferation, survival and dependent cell lines treated with eFT226. metastasis Conclusions TSC ERK eFT226 Treatment Inhibits RTK Expression In vivo • RTK signaling activates pathways • eFT226 is a potent and selective inhibitor of eIF4A1 dependent B. C. amp that enhance eIF4A activity SNU-16 MDA-MB-361 A. NCI-H716 (FGFR2 ) B. translation mTOR wt FGFR2 (∆UTR) wt HER2 (∆UTR) • eFFECTOR Therapeutics has designed Vehicle eFT226 (1mg/kg) eFT226 (nM) eFT226 (nM) eFT226 (nM) eFT226 (nM) FGFR2 • eFT226 translationally down regulates oncogenic RTK protein 0 25 100 0 25 100 0 30 100 0 30 100 eFT226, a potent, small molecule p-Akt S473 expression (e.g., FGFR1/2 and HER2) through the formation of a S6K1 RSK FGFR2 HER2 sequence selective inhibitor of eIF4A Akt stable ternary complex [eIF4A-eFT226-mRNA] with specific 5’-UTR Cyclin D1 Cyclin D1 mediated translation Tubulin Degradation Tubulin Vinculin motifs P P C. NCI-H1581 (FGFR1amp) D. eIF4B PDCD4 • eFT226 inhibits tumor cell proliferation and induces apoptosis Figure 2. Protein expression of RTKs are regulated by eFT226 through the 5’-UTR. A) Luciferase reporter eFT226 gene constructs containing 5’-UTRs of FGFR1/2, HER2 or TUBA were transiently transfected into the Vehicle 0.3 mg/kg 1 mg/kg • eFT226 is well-tolerated and shows significant in vivo tumor growth HEK293t cells and treated with increasing concentrations of eFT226 for 4 hr. B-C) Predicted eFT226 FGFR1 inhibition and regression in RTK driven xenograft models binding sites were deleted from 5’-UTR (∆UTR) constructs.
Recommended publications
  • A Computational Approach for Defining a Signature of Β-Cell Golgi Stress in Diabetes Mellitus
    Page 1 of 781 Diabetes A Computational Approach for Defining a Signature of β-Cell Golgi Stress in Diabetes Mellitus Robert N. Bone1,6,7, Olufunmilola Oyebamiji2, Sayali Talware2, Sharmila Selvaraj2, Preethi Krishnan3,6, Farooq Syed1,6,7, Huanmei Wu2, Carmella Evans-Molina 1,3,4,5,6,7,8* Departments of 1Pediatrics, 3Medicine, 4Anatomy, Cell Biology & Physiology, 5Biochemistry & Molecular Biology, the 6Center for Diabetes & Metabolic Diseases, and the 7Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202; 2Department of BioHealth Informatics, Indiana University-Purdue University Indianapolis, Indianapolis, IN, 46202; 8Roudebush VA Medical Center, Indianapolis, IN 46202. *Corresponding Author(s): Carmella Evans-Molina, MD, PhD ([email protected]) Indiana University School of Medicine, 635 Barnhill Drive, MS 2031A, Indianapolis, IN 46202, Telephone: (317) 274-4145, Fax (317) 274-4107 Running Title: Golgi Stress Response in Diabetes Word Count: 4358 Number of Figures: 6 Keywords: Golgi apparatus stress, Islets, β cell, Type 1 diabetes, Type 2 diabetes 1 Diabetes Publish Ahead of Print, published online August 20, 2020 Diabetes Page 2 of 781 ABSTRACT The Golgi apparatus (GA) is an important site of insulin processing and granule maturation, but whether GA organelle dysfunction and GA stress are present in the diabetic β-cell has not been tested. We utilized an informatics-based approach to develop a transcriptional signature of β-cell GA stress using existing RNA sequencing and microarray datasets generated using human islets from donors with diabetes and islets where type 1(T1D) and type 2 diabetes (T2D) had been modeled ex vivo. To narrow our results to GA-specific genes, we applied a filter set of 1,030 genes accepted as GA associated.
    [Show full text]
  • A Helicase-Independent Activity of Eif4a in Promoting Mrna Recruitment to the Human Ribosome
    A helicase-independent activity of eIF4A in promoting mRNA recruitment to the human ribosome Masaaki Sokabea and Christopher S. Frasera,1 aDepartment of Molecular and Cellular Biology, College of Biological Sciences, University of California, Davis, CA 95616 Edited by Alan G. Hinnebusch, National Institutes of Health, Bethesda, MD, and approved May 5, 2017 (received for review December 12, 2016) In the scanning model of translation initiation, the decoding site and at the solvent side of the mRNA entry channel (14). Importantly, latch of the 40S subunit must open to allow the recruitment and that study showed that a short mRNA that does not extend into the migration of messenger RNA (mRNA); however, the precise molec- entry channel fails to displace eIF3j. A similar observation was also ular details for how initiation factors regulate mRNA accommodation found for initiation mediated by the hepatitis C virus internal ribo- into the decoding site have not yet been elucidated. Eukaryotic some entry site, where an mRNA truncated after the initiation co- initiation factor (eIF) 3j is a subunit of eIF3 that binds to the mRNA don failed to displace eIF3j (11). Taken together, these studies entry channel and A-site of the 40S subunit. Previous studies have suggest a model in which a full accommodation of mRNA in the shown that a reduced affinity of eIF3j for the 43S preinitiation mRNA entry channel of the 40S subunit corresponds to a reduced complex (PIC) occurs on eIF4F-dependent mRNA recruitment. Because affinity of eIF3j for the 40S subunit. This model has allowed us to eIF3j and mRNA bind anticooperatively to the 43S PIC, reduced eIF3j exploit the change in eIF3j affinity for the 43S PIC to quantitatively affinity likely reflects a state of full accommodation of mRNA into the monitor the process of mRNA recruitment.
    [Show full text]
  • Ten Commandments for a Good Scientist
    Unravelling the mechanism of differential biological responses induced by food-borne xeno- and phyto-estrogenic compounds Ana María Sotoca Covaleda Wageningen 2010 Thesis committee Thesis supervisors Prof. dr. ir. Ivonne M.C.M. Rietjens Professor of Toxicology Wageningen University Prof. dr. Albertinka J. Murk Personal chair at the sub-department of Toxicology Wageningen University Thesis co-supervisor Dr. ir. Jacques J.M. Vervoort Associate professor at the Laboratory of Biochemistry Wageningen University Other members Prof. dr. Michael R. Muller, Wageningen University Prof. dr. ir. Huub F.J. Savelkoul, Wageningen University Prof. dr. Everardus J. van Zoelen, Radboud University Nijmegen Dr. ir. Toine F.H. Bovee, RIKILT, Wageningen This research was conducted under the auspices of the Graduate School VLAG Unravelling the mechanism of differential biological responses induced by food-borne xeno- and phyto-estrogenic compounds Ana María Sotoca Covaleda Thesis submitted in fulfillment of the requirements for the degree of doctor at Wageningen University by the authority of the Rector Magnificus Prof. dr. M.J. Kropff, in the presence of the Thesis Committee appointed by the Academic Board to be defended in public on Tuesday 14 September 2010 at 4 p.m. in the Aula Unravelling the mechanism of differential biological responses induced by food-borne xeno- and phyto-estrogenic compounds. Ana María Sotoca Covaleda Thesis Wageningen University, Wageningen, The Netherlands, 2010, With references, and with summary in Dutch. ISBN: 978-90-8585-707-5 “Caminante no hay camino, se hace camino al andar. Al andar se hace camino, y al volver la vista atrás se ve la senda que nunca se ha de volver a pisar” - Antonio Machado – A mi madre.
    [Show full text]
  • Initiation Factor Eif5b Catalyzes Second GTP-Dependent Step in Eukaryotic Translation Initiation
    Initiation factor eIF5B catalyzes second GTP-dependent step in eukaryotic translation initiation Joon H. Lee*†, Tatyana V. Pestova†‡§, Byung-Sik Shin*, Chune Cao*, Sang K. Choi*, and Thomas E. Dever*¶ *Laboratory of Gene Regulation and Development, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892-2716; ‡Department of Microbiology and Immunology, State University of New York Health Science Center, Brooklyn, NY 11203; and §A. N. Belozersky Institute of Physico-Chemical Biology, Moscow State University, Moscow, Russia Edited by Harry F. Noller, University of California, Santa Cruz, CA, and approved October 31, 2002 (received for review September 19, 2002) Initiation factors IF2 in bacteria and eIF2 in eukaryotes are GTPases In addition, when nonhydrolyzable GDPNP was substituted Met that bind Met-tRNAi to the small ribosomal subunit. eIF5B, the for GTP, eIF5B catalyzed subunit joining; however, the factor eukaryotic ortholog of IF2, is a GTPase that promotes ribosomal was unable to dissociate from the 80S ribosome after subunit subunit joining. Here we show that eIF5B GTPase activity is re- joining (7). quired for protein synthesis. Mutation of the conserved Asp-759 in To dissect the function of the eIF5B G domain and test the human eIF5B GTP-binding domain to Asn converts eIF5B to an model that two GTP molecules are required in translation XTPase and introduces an XTP requirement for subunit joining and initiation, we mutated conserved residues in the eIF5B G translation initiation. Thus, in contrast to bacteria where the single domain and tested the function of the mutant proteins in GTPase IF2 is sufficient to catalyze translation initiation, eukaryotic translation initiation.
    [Show full text]
  • Eif4a Is Stimulated by the Pre-Initiation Complex and Enhances Recruitment of Mrnas Regardless of Structural Complexity
    bioRxiv preprint doi: https://doi.org/10.1101/147959; this version posted June 13, 2017. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license. 1 eIF4A is stimulated by the pre-initiation complex and enhances recruitment of mRNAs regardless of structural 2 complexity 3 Paul Yourik1, Colin Echeverría Aitken1, Fujun Zhou1, Neha Gupta1,2, Alan G. Hinnebusch2,3, Jon R. Lorsch1,3 4 5 1Laboratory on the Mechanism and Regulation of Protein Synthesis, Eunice Kennedy Shriver National Institute of Child 6 Health and Development, National Institutes of Health, Bethesda, MD 20892 7 2Laboratory of Gene Regulation and Development, Eunice Kennedy Shriver National Institute of Child Health and 8 Human Development, National Institutes of Health, Bethesda, MD 20892, USA 9 3Corresponding Author 10 11 ABSTRACT 12 eIF4A is a DEAD-box RNA-dependent ATPase thought to unwind RNA secondary structure in the 5'-untranslated 13 regions (UTRs) of mRNAs to promote their recruitment to the eukaryotic translation pre-initiation complex (PIC). We 14 show that the PIC stimulates the ATPase of eIF4A, indicating that the factor acts in association with initiating ribosomal 15 complexes rather than exclusively on isolated mRNAs. ATP hydrolysis by eIF4A accelerates the rate of recruitment for 16 all mRNAs tested, regardless of their degree of secondary structure, indicating that the factor plays important roles 17 beyond unwinding mRNA structure. Structures in the 5'-UTR and 3' of the start codon synergistically inhibit mRNA 18 recruitment, in a manner relieved by eIF4A, suggesting that the factor resolves global mRNA structure rather than just 19 secondary structures in the 5'-UTR.
    [Show full text]
  • Whole Exome Sequencing in Families at High Risk for Hodgkin Lymphoma: Identification of a Predisposing Mutation in the KDR Gene
    Hodgkin Lymphoma SUPPLEMENTARY APPENDIX Whole exome sequencing in families at high risk for Hodgkin lymphoma: identification of a predisposing mutation in the KDR gene Melissa Rotunno, 1 Mary L. McMaster, 1 Joseph Boland, 2 Sara Bass, 2 Xijun Zhang, 2 Laurie Burdett, 2 Belynda Hicks, 2 Sarangan Ravichandran, 3 Brian T. Luke, 3 Meredith Yeager, 2 Laura Fontaine, 4 Paula L. Hyland, 1 Alisa M. Goldstein, 1 NCI DCEG Cancer Sequencing Working Group, NCI DCEG Cancer Genomics Research Laboratory, Stephen J. Chanock, 5 Neil E. Caporaso, 1 Margaret A. Tucker, 6 and Lynn R. Goldin 1 1Genetic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Bethesda, MD; 2Cancer Genomics Research Laboratory, Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Bethesda, MD; 3Ad - vanced Biomedical Computing Center, Leidos Biomedical Research Inc.; Frederick National Laboratory for Cancer Research, Frederick, MD; 4Westat, Inc., Rockville MD; 5Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Bethesda, MD; and 6Human Genetics Program, Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Bethesda, MD, USA ©2016 Ferrata Storti Foundation. This is an open-access paper. doi:10.3324/haematol.2015.135475 Received: August 19, 2015. Accepted: January 7, 2016. Pre-published: June 13, 2016. Correspondence: [email protected] Supplemental Author Information: NCI DCEG Cancer Sequencing Working Group: Mark H. Greene, Allan Hildesheim, Nan Hu, Maria Theresa Landi, Jennifer Loud, Phuong Mai, Lisa Mirabello, Lindsay Morton, Dilys Parry, Anand Pathak, Douglas R. Stewart, Philip R. Taylor, Geoffrey S. Tobias, Xiaohong R. Yang, Guoqin Yu NCI DCEG Cancer Genomics Research Laboratory: Salma Chowdhury, Michael Cullen, Casey Dagnall, Herbert Higson, Amy A.
    [Show full text]
  • Supplementary Material Contents
    Supplementary Material Contents Immune modulating proteins identified from exosomal samples.....................................................................2 Figure S1: Overlap between exosomal and soluble proteomes.................................................................................... 4 Bacterial strains:..............................................................................................................................................4 Figure S2: Variability between subjects of effects of exosomes on BL21-lux growth.................................................... 5 Figure S3: Early effects of exosomes on growth of BL21 E. coli .................................................................................... 5 Figure S4: Exosomal Lysis............................................................................................................................................ 6 Figure S5: Effect of pH on exosomal action.................................................................................................................. 7 Figure S6: Effect of exosomes on growth of UPEC (pH = 6.5) suspended in exosome-depleted urine supernatant ....... 8 Effective exosomal concentration....................................................................................................................8 Figure S7: Sample constitution for luminometry experiments..................................................................................... 8 Figure S8: Determining effective concentration .........................................................................................................
    [Show full text]
  • Rps3/Us3 Promotes Mrna Binding at the 40S Ribosome Entry Channel and Stabilizes Preinitiation Complexes at Start Codons
    Rps3/uS3 promotes mRNA binding at the 40S ribosome entry channel and stabilizes preinitiation complexes at start codons Jinsheng Donga, Colin Echeverría Aitkenb, Anil Thakura, Byung-Sik Shina, Jon R. Lorschb,1, and Alan G. Hinnebuscha,1 aLaboratory of Gene Regulation and Development, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892; and bLaboratory on the Mechanism and Regulation of Protein Synthesis, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892 Contributed by Alan G. Hinnebusch, January 24, 2017 (sent for review December 15, 2016; reviewed by Jamie H. D. Cate and Matthew S. Sachs) Met The eukaryotic 43S preinitiation complex (PIC) bearing Met-tRNAi rearrangement to PIN at both near-cognate start codons (e.g., in a ternary complex (TC) with eukaryotic initiation factor (eIF)2-GTP UUG) and cognate (AUG) codons in poor Kozak context; hence scans the mRNA leader for an AUG codon in favorable “Kozak” eIF1 must dissociate from the 40S subunit for start-codon rec- context. AUG recognition provokes rearrangement from an open ognition (Fig. 1A). Consistent with this, structural analyses of PIC conformation with TC bound in a state not fully engaged with partial PICs reveal that eIF1 and eIF1A promote rotation of the “ ” the P site ( POUT ) to a closed, arrested conformation with TC tightly 40S head relative to the body (2, 3), thought to be instrumental bound in the “P ” state. Yeast ribosomal protein Rps3/uS3 resides IN in TC binding in the POUT conformation, but that eIF1 physically in the mRNA entry channel of the 40S subunit and contacts mRNA Met clashes with Met-tRNAi in the PIN state (2, 4), and is both via conserved residues whose functional importance was unknown.
    [Show full text]
  • Relevance of Translation Initiation in Diffuse Glioma Biology and Its
    cells Review Relevance of Translation Initiation in Diffuse Glioma Biology and its Therapeutic Potential Digregorio Marina 1, Lombard Arnaud 1,2, Lumapat Paul Noel 1, Scholtes Felix 1,2, Rogister Bernard 1,3 and Coppieters Natacha 1,* 1 Laboratory of Nervous System Disorders and Therapy, GIGA-Neurosciences Research Centre, University of Liège, 4000 Liège, Belgium; [email protected] (D.M.); [email protected] (L.A.); [email protected] (L.P.N.); [email protected] (S.F.); [email protected] (R.B.) 2 Department of Neurosurgery, CHU of Liège, 4000 Liège, Belgium 3 Department of Neurology, CHU of Liège, 4000 Liège, Belgium * Correspondence: [email protected] Received: 18 October 2019; Accepted: 26 November 2019; Published: 29 November 2019 Abstract: Cancer cells are continually exposed to environmental stressors forcing them to adapt their protein production to survive. The translational machinery can be recruited by malignant cells to synthesize proteins required to promote their survival, even in times of high physiological and pathological stress. This phenomenon has been described in several cancers including in gliomas. Abnormal regulation of translation has encouraged the development of new therapeutics targeting the protein synthesis pathway. This approach could be meaningful for glioma given the fact that the median survival following diagnosis of the highest grade of glioma remains short despite current therapy. The identification of new targets for the development of novel therapeutics is therefore needed in order to improve this devastating overall survival rate. This review discusses current literature on translation in gliomas with a focus on the initiation step covering both the cap-dependent and cap-independent modes of initiation.
    [Show full text]
  • The Mtorc1-4E-BP-Eif4e Axis Controls De Novo Bcl6 Protein Synthesis in T Cells and Systemic Autoimmunity
    ARTICLE DOI: 10.1038/s41467-017-00348-3 OPEN The mTORC1-4E-BP-eIF4E axis controls de novo Bcl6 protein synthesis in T cells and systemic autoimmunity Woelsung Yi1, Sanjay Gupta1, Edd Ricker2, Michela Manni1, Rolf Jessberger3, Yurii Chinenov4,5, Henrik Molina6 & Alessandra B. Pernis1,2,7 Post-transcriptional modifications can control protein abundance, but the extent to which these alterations contribute to the expression of T helper (TH) lineage-defining factors is unknown. Tight regulation of Bcl6 expression, an essential transcription factor for T follicular helper (TFH) cells, is critical as aberrant TFH cell expansion is associated with autoimmune diseases, such as systemic lupus erythematosus (SLE). Here we show that lack of the SLE risk variant Def6 results in deregulation of Bcl6 protein synthesis in T cells as a result of enhanced activation of the mTORC1–4E-BP–eIF4E axis, secondary to aberrant assembly of a raptor–p62–TRAF6 complex. Proteomic analysis reveals that this pathway selectively controls the abundance of a subset of proteins. Rapamycin or raptor deletion ameliorates the aberrant TFH cell expansion in mice lacking Def6. Thus deregulation of mTORC1-dependent pathways controlling protein synthesis can result in T-cell dysfunction, indicating a mechanism by which mTORC1 can promote autoimmunity. 1 Autoimmunity and Inflammation Program, Hospital for Special Surgery, 535 East 70th Street, New York, New York 10021 USA. 2 Graduate Program in Immunology and Microbial Pathogenesis, Weill Cornell Graduate School of Medical Sciences, 1300 York Avenue, Box 65, New York, New York 10021 USA. 3 Institute of Physiological Chemistry, Technische Universität Dresden, Fiedlerstrasse 42, MTZ, 01307 Dresden, Germany.
    [Show full text]
  • Loss of Eif4e Phosphorylation Engenders Depression-Like Behaviors Via Selective Mrna Translation
    This Accepted Manuscript has not been copyedited and formatted. The final version may differ from this version. A link to any extended data will be provided when the final version is posted online. Research Articles: Neurobiology of Disease Loss of eIF4E phosphorylation engenders depression-like behaviors via selective mRNA translation Inês Silva Amorim1,2, Sonal Kedia1,2, Stella Kouloulia1,2, Konstanze Simbriger1,2, Ilse Gantois3, Seyed Mehdi Jafarnejad3, Yupeng Li1,2, Agniete Kampaite1,2, Tine Pooters1, Nicola Romanò1 and Christos G. Gkogkas1,2,4 1Centre for Discovery Brain Sciences, EH8 9XD, Edinburgh, Scotland, UK 2The Patrick Wild Centre, EH8 9XD, Edinburgh, Scotland, UK 3Goodman Cancer Research Centre and Biochemistry Department, McGill University, H3A 1A3, Montréal, QC, Canada 4Simons Initiative for the Developing Brain, EH8 9XD, Edinburgh, Scotland, UK DOI: 10.1523/JNEUROSCI.2673-17.2018 Received: 16 September 2017 Revised: 3 December 2017 Accepted: 8 January 2018 Published: 24 January 2018 Author contributions: I.S.A., S. Kedia, S. Kouloulia, S.M.J., N.R., and C.G.G. designed research; I.S.A., S. Kedia, S. Kouloulia, K.S., I.G., S.M.J., Y.L., A.K., N.R., and C.G.G. performed research; I.S.A., S. Kedia, S. Kouloulia, K.S., I.G., S.M.J., A.K., T.P., and C.G.G. analyzed data; N.R. and C.G.G. contributed unpublished reagents/analytic tools; C.G.G. wrote the paper. Conflict of Interest: The authors declare no competing financial interests. This work was supported by grants to CGG: Sir Henry Dale Fellowship from the Wellcome Trust and Royal Society (107687/Z/15/Z), a NARSAD Young Investigator grant from the Brain & Behavior Research Foundation, the RS Macdonald Charitable Trust and the Patrick Wild Centre.
    [Show full text]
  • Inhibition of the MID1 Protein Complex
    Matthes et al. Cell Death Discovery (2018) 4:4 DOI 10.1038/s41420-017-0003-8 Cell Death Discovery ARTICLE Open Access Inhibition of the MID1 protein complex: a novel approach targeting APP protein synthesis Frank Matthes1,MoritzM.Hettich1, Judith Schilling1, Diana Flores-Dominguez1, Nelli Blank1, Thomas Wiglenda2, Alexander Buntru2,HannaWolf1, Stephanie Weber1,InaVorberg 1, Alina Dagane2, Gunnar Dittmar2,3,ErichWanker2, Dan Ehninger1 and Sybille Krauss1 Abstract Alzheimer’s disease (AD) is characterized by two neuropathological hallmarks: senile plaques, which are composed of amyloid-β (Aβ) peptides, and neurofibrillary tangles, which are composed of hyperphosphorylated tau protein. Aβ peptides are derived from sequential proteolytic cleavage of the amyloid precursor protein (APP). In this study, we identified a so far unknown mode of regulation of APP protein synthesis involving the MID1 protein complex: MID1 binds to and regulates the translation of APP mRNA. The underlying mode of action of MID1 involves the mTOR pathway. Thus, inhibition of the MID1 complex reduces the APP protein level in cultures of primary neurons. Based on this, we used one compound that we discovered previously to interfere with the MID1 complex, metformin, for in vivo experiments. Indeed, long-term treatment with metformin decreased APP protein expression levels and consequently Aβ in an AD mouse model. Importantly, we have initiated the metformin treatment late in life, at a time-point where mice were in an already progressed state of the disease, and could observe an improved behavioral phenotype. These 1234567890 1234567890 findings together with our previous observation, showing that inhibition of the MID1 complex by metformin also decreases tau phosphorylation, make the MID1 complex a particularly interesting drug target for treating AD.
    [Show full text]